eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
5/2014
vol. 18
 
Share:
Share:
Original paper

The ability of hyaluronan fragments to reverse the resistance of C6 rat glioma cell line to temozolomide and carmustine

Michał Seweryn Karbownik
,
Tadeusz Pietras
,
Janusz Szemraj
,
Edward Kowalczyk
,
Jerzy Zygmunt Nowak

Contemp Oncol (Pozn) 2014; 18 (5): 323–328
Online publish date: 2014/09/23
Article file
- The ability.pdf  [0.09 MB]
Get citation
 
PlumX metrics:
 

Introduction

Resistance to anti-cancer drugs

In recent decades, significant progress has been made in understanding cancer biology and inventing new therapeutic strategies. Despite these achievements, we are still far from successful management of cancer. Resistance to anti-cancer therapies, both intrinsic and acquired, constitutes one of the main causes of an inadequate response to treatment and diminished survival of cancer patients [1–3]. Drug resistance arises in a wide variety of mechanisms. It may develop due to decreased uptake of a drug into a cancer cell [4, 5], enhanced efflux of a drug out of a cancer cell, which is commonly mediated by ATP-binding cassette (ABC) transporters [6, 7], decreased activation of an anti-cancer pro-drug or inactivation of an active drug [8, 9], altered CYP-mediated metabolism [10], mutation or amplification of the targeted protein [11, 12], activation of alternative survival pathways [13], activation of repair mechanisms [14], and increased activity of anti-apoptotic signals [15, 16].
Resistance to anti-cancer drugs is primarily initiated by genetic factors [17]. However, the environment-mediated drug resistance model (EMDR) has been proposed as an alternative hypothesis explaining the origin of cancer cell resistance [18] – these hypotheses are not mutually exclusive. According to the EMDR model, resistance emerges as a result of an intimate relationship and subsequent signaling dialogue between cancer cells and the surrounding microenvironment [19, 20]. EMDR is mediated by either soluble factors present in the tumor microenvironment or cell adhesion-related factors [19]. Soluble factor-mediated drug resistance (SFM-DR) occurs through cytokines, chemokines and growth factors secreted by neighboring fibroblast-like tumor stroma [21, 22]. Cell adhesion-mediated drug resistance (CAM-DR) results from adhesion of tumor cell integrins to stromal fibroblasts [23] or to components of the extracellular matrix (ECM) [24].

Hyaluronan and drug resistance

One of the main components of the extracellular matrix is hyaluronan (HA). It is a linear high-molecular-weight polymer composed of repeating units of D-glucuronic acid residues and N-acetyl-D-glucosamine numbering up to 25,000 disaccharide units, which gives a molecular mass of up to 10 MDa. Hyaluronan is distributed ubiquitously in human tissues. It abundantly occurs in the vitreous body of the eye, synovial fluid, and connective tissue. Over half of the total body hyaluronan is present in the skin. Hyaluronan clearly plays a structural role in the organism. It maintains appropriate tissue hydration and tension, provides necessary lubrication in joint cavities, and decreases friction in tendon sheaths [25–27].
Hyaluronan-evoked anti-cancer drug resistance may be of a physico-mechanical nature as a dense extracellular matrix limits the delivery and distribution of therapeutic agents [28]. That is why enzymatic depletion of HA may be explored as a means to improve drug delivery [29].
Although hyaluronan has been perceived for decades as being only a passive component of the tissues [30], now it is evident that HA also mediates various intracellular signaling pathways involved in embryonic morphogenesis [31], inflammation and immune regulation [32], wound repair [33], and cancer [34]. Hyaluronan exerts its function through the interaction with cell surface receptors. CD44 is a transmembrane protein that constitutes a major hyaluronan-binding receptor [35–37].
CD44 (mainly its alternative splice variants) has been observed to be up-regulated in many cancers, including cancer stem-like cells, and to correlate with malignant phenotype of the cells [37–41]. The relevance of hyaluronan-CD44 interaction in environment-mediated drug resistance has been explicitly confirmed in many studies [42–45]. Overproduction of hyaluronan stimulates drug resistance in drug-sensitive cancer cells [46], and the phenomenon is likely to be mediated in part through the apoptotic pathways [47]. However, the regulation of drug transporters, including P-glycoprotein (also known as multidrug resistance protein 1; P-gp/MDR1 encoded by the gene ABCB1) [48], multidrug resistance-associated protein 2 (MRP2 encoded by the gene ABCC2) [48, 49] and breast cancer resistance protein (ABC family drug transporter BCRP encoded by the gene ABCG2) [50] is a major contributor to HA-CD44-mediated drug resistance. Interestingly, P-gp has also been found to be localized in close molecular vicinity and to be functionally associated with CD44 [24, 51].

Tackling drug resistance

Research efforts in cancer biology and pharmacology are nowadays focused on developing therapeutic strategies to overcome resistance to anti-cancer drugs [7, 18, 52, 53]. Hyaluronan-CD44-dependent drug resistance may be defeated by disrupting HA-CD44 interaction. It may be achieved by using anti-CD44 antibodies, small interfering RNA (siRNA) or short hairpin RNA (shRNA), soluble HA-binding proteins, and small hyaluronan oligosaccharides (oHA) [24, 37].
Attempts to use small hyaluronan oligosaccharides as agents reversing resistance to anti-cancer drugs are being successfully made [50, 54–58]. As CD44 receptor co-localizes with P-glycoprotein, treating the cells with hyaluronan oligomers induces rapid internalization of both the efflux pump and CD44 into the cell [24]. Moreover, disruption of endogenous hyaluronan-induced signaling in mammary carcinoma cells using small hyaluronan oligomers (3–9 disaccharide units) suppresses resistance to vincristine by 10-fold, to paclitaxel by 12-fold, and to carmustine by 78-fold. It also reverses resistance to doxorubicin and methotrexate [46, 56].
Similar effects have been reported in malignant glioma models. The addition of the oHA fraction (3–10 disaccharide units) to methotrexate-resistant glioma cell culture significantly reduced cell viability in the presence of methotrexate. In vivo, the glioma growth rate was suppressed by oHA, possibly by decreasing recruitment of host-derived BCRP-positive progenitor cells into the engrafted gliomas [50]. CD44 depletion by shRNA-mediated knockdown also resulted in glioma cell lines’ sensitization to temozolomide and carmustine, the first-line cytotoxic drugs for glioblastoma multiforme. However, in that study, HA oligomers were not tested [38].

Aim of the study

The findings presented above suggest that hyaluronan oligomers may reverse drug resistance of glioma cells. However, the reversal effects of oHA of defined molecular length on glioma cells have not been investigated yet. In this study, we examined three hyaluronan fragments – oligomers containing 2 disaccharide units, 5 disaccharide units, and hyaluronan of the weight of 68 (±5%) kDa (approximately 180 disaccharide units) – as agents possibly capable of reversing the resistance of a C6 rat glioma cell line to temozolomide and carmustine.

Material and methods

Reagents

3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), temozolomide (TMZ) and carmustine (BCNU) were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Hyaluronan fragments – oligomers containing 2 disaccharide units (oHA-2), 5 disaccharide units (oHA-5), and hyaluronan of the weight of 68 (±5%) kDa (HA-68k) – were purchased from Lifecore Biomedical (Chaska, MN, USA).

Cell culture

The rat C6 glioma cell line was obtained from the European Collection of Animal Cell Cultures (Porton Down, UK). Dulbecco’s Modified Eagle’s Medium with 1,000 mg/l glucose (DMEM), fetal bovine serum (FBS), penicillin-streptomycin solution (10,000 units/ml penicillin and 10 mg/ml streptomycin in normal saline), phosphate buffered saline (PBS; pH 7.4) and trypsin-EDTA (0.25% trypsin, 1 mM EDTA-4 Na) were purchased from Invitrogen (Carlsbad, CA, USA). L-glutamine was purchased from Sigma Chemical Co. (St. Louis, MO, USA). C6 glioma cells were grown in 60-mm Petri dishes in DMEM with 1,000 mg/l glucose, supplemented with 10% FBS, L-glutamine (2 mM), penicillin (100 units/ml), and streptomycin (100 µg/ml). The cells were maintained at 37°C in humidified atmosphere of 95% air and 5% CO2. For subcultures, cells were harvested in trypsin–EDTA solution twice a week and seeded at a density of 1.0 × 106 cells per dish.

Cell viability assay

Cell viability and mitochondrial function were measured by MTT reduction to formazan derivative through cellular mitochondrial dehydrogenases. C6 glioma cells were seeded at a density of 1.5 × 104 cells per well in 96-well plates and grown in standard culture conditions for twenty-four hours. Then, the culture medium was replaced with fresh serum-free medium and the cells were exposed to the tested chemicals for the next twenty-four hours.
Firstly, hyaluronan fragments were dissolved in sterile water. The cells were exposed to HA fragments of 10, 50, and 150 µg/ml to ensure their non-toxicity. Secondly, TMZ was dissolved in dimethyl sulfoxide (DMSO), and BCNU was dissolved in ethanol 96% to produce the final concentrations of 150 mM. The cells were exposed to TMZ of 125, 250, 500, 750, 1,000, and 1,500 µM, and BCNU of 31.25, 62.5, 125, 250, 500, and 1,000 µM in order to plot the calibration curves and to determine IC50 values. The cells were also exposed to solvents of TMZ and BCNU in corresponding concentrations to ensure their non-toxicity. Finally, in order to investigate the reversal effects of HA fragments on TMZ and BCNU resistance, the cells were treated with 500 µM of TMZ or 125 µM of BCNU in the presence or absence of hyaluronan fragments of 50 µg/ml.
After incubation for twenty-four hours, MTT in the final concentration of 1.0 mg/ml was added and the cells were incubated for 4 h at 37°C. The supernatants were carefully aspirated. Formazan crystals were solubilized in DMSO and absorbance, directly proportional to the number of viable cells, was measured at 570 nm using a microplate reader (BioTek, EL ×800). The results were expressed as the relative absorbance, i.e. (A570 of experimental wells) / (A570 of control wells) × 100%.

Statistical analysis

The normal distribution of parameters was confirmed by the Shapiro-Wilks test; therefore data were described as the mean ± SD (standard deviation), and analyzed by Student’s t-test. P-values below 0.05 were considered statistically significant. The analysis was performed using STATISTICA 10 StatSoft software.

Results

Effects of HA fragments on the growth of C6 glioma cells We did not find any statistically significant differences between the viability of the cells treated with oHA-2, oHA-5, and HA-68k in the concentration range of 10–150 µg/ml and the control wells. The relative absorbance in the viability assay is presented in Table 1. The results suggest that none of the HA fragments had any inhibitory effects on the growth of C6 glioma cells.

Inhibitory effects of TMZ and BCNU on the growth of C6 cells

The solvents of TMZ and BCNU in corresponding concentrations, i.e. DMSO in the concentration range of 0.083–1.0% and ethanol in the concentration range of 0.021–0.67%, respectively, had no inhibitory effects on the growth of C6 glioma cells. Treatment of C6 cells with 125, 250, 500, 750, 1,000, and 1,500 µM of TMZ produced the relative absorbance in MTT viability assay of 96.9, 84.9, 60.4, 48.6, 39.4, and 33.4, respectively (Fig. 1), while treatment of C6 cells with 31.25, 62.5, 125, 250, 500, and 1,000 µM of BCNU produced the relative absorbance of 87.6, 74.0, 62.4, 45.3, 24.7, and 2.4, respectively (Fig. 2). The regression equation of the inhibitory effect of TMZ was y = –27.42 × ln(x) + 231.56 (r = 0.9934), and that of BCNU was y = –24.35 × ln(x) + 175.39 (r = 0.9922), with the IC50 = 751 µM for TMZ, and 172 µM for BCNU.

Reversal effects of HA fragments on TMZ and BCNU resistance in C6 glioma cells

The C6 cells were incubated with 500 µM of TMZ or 125 µM of BCNU in the presence or absence of hyaluronan fragments of 50 µg/ml. In the MTT assay statistically significant decreases in the viability of cells occurred in the presence of TMZ+oHA-5 compared to TMZ alone (51.2 ±4.5 vs. 74.2 ±5.8, p = 0.0031), BCNU+o-HA5 compared to BCNU alone (49.3 ±4.4 vs. 65.6 ±5.7, p = 0.0119), and BCNU+HA-68k compared to BCNU alone (55.2 ±2.3 vs. 65.6 ±5.7, p = 0.0496). Data are presented in Fig. 3.

Discussion

Malignant gliomas, including particularly glioblastoma multiforme (GBM), are highly challenging to treat. Surgery is the first therapeutic approach to GBM, but infiltrative characteristics of the tumor make complete resection virtually impossible. Radiation therapy (RT) is the second mainstay of GBM treatment. RT is frequently combined with adjuvant chemotherapy, as it results in a survival benefit with minimal additional toxicity [59]. Most treatment protocols employ alkylating agents: temozolomide (TMZ), or nitrosoureas, e.g. carmustine (BCNU) [60].
Gliomas are known for their resistance to therapy [61]. The objective response rates of GBM to chemotherapy (except oligodendroglial subtypes) are approximately 30%, and time to progression (TTP) is short (3–6 months) [62]. The mechanisms of resistance to TMZ and BCNU in malignant gliomas are mediated by: (1) hyperactivity of methylguanine methyltransferase (MGMT) repair enzyme, which removes alkyl groups from the O6 position of guanine, (2) defects in the mismatch repair system (MMR), which makes the cells tolerant to mispairing of O6-methylguanine to thymine and stops apoptosis, (3) poly(ADP-ribose)polymerase (PARP) activity, which is involved in DNA adduct repair and contributes to cell survival, (4) dysregulation of apoptosis-regulating genes and the proteins Bcl-2, Bcl-XL, p53, and EGFR [63], or (5) ATP-binding cassette transporters’ overexpression [64].
Small hyaluronan oligomers have been widely investigated as agents potentially reversing resistance to anti-cancer drugs. However, there is little evidence of effects of oHA of defined molecular length on reversing glioma resistance to alkylating agents. As it has been reported that HA degradation products had extremely different biological functions [37], we investigated HA fragments of various lengths: oligomers containing 2 disaccharide units (oHA-2), 5 disaccharide units (oHA-5), and hyaluronan of the weight of 68 (±5%) kDa (HA-68k), i.e. containing approximately 180 disaccharide units. The results showed that addition of oHA-5 to TMZ or BCNU treatment of the cells significantly reduced cell viability, effectively reversing both TMZ and BCNU resistance. No significant effect of TMZ+oHA-2 vs. TMZ alone, BCNU+oHA-2 vs. BCNU alone, or TMZ+HA-68k vs. TMZ alone on cell viability was found. The significance of the effect of HA-68k on BCNU resistance is uncertain, as the p-value approaches 0.05.
The positive effect of oHA-5 and non-significant effect of HA-68k on reversing resistance to TMZ and BCNU are in accordance with pharmacodynamic analysis of the quality of HA-CD44 interaction, assuming the reversal effect is CD44-mediated [38]. The analysis revealed that HA oligomers of 3–9 disaccharide units (including oHA-5), because of their shortness, exhibit only monovalent binding to CD44, displacing any hyaluronan polymer from membrane-bound receptors, serving as receptor antagonists. At approximately 10–19 disaccharide units, a progressive 3-fold increase in avidity was seen, suggesting that divalent binding and subsequent intracellular signaling occur. Larger polymers (including HA-68k) also induce CD44-mediated signaling [37, 58, 65].
The non-significant impact of the smallest hyaluronan oligomer, oHA-2, on anti-cancer drug resistance is not consistent with the research by Cui et al. [57]. They suggested that the smaller the oHA, the stronger the reversal effect is. The lack of statistical significance may be due to inadequate oHA concentration, as Cui et al. reached the significance at oHA concentration of ≥ 100 µg/ml. On the other hand, oHA-2 fragments may produce a different effect than larger oligomers, as neither Misra et al. [46] Gilg et al. [50] nor Lesley et al. [65] included oHA-2 in their analyses.
To assess the reversal effects of hyaluronan fragments, TMZ and BCNU were used at concentrations of 500 µM and 125 µM, respectively. Concentrations somewhat lower than the IC50 values were deliberately chosen as it is unlikely that concentrations as high as IC50 values are reached in brain tissues during chemotherapeutic drug treatment in the clinical setting. Patients with brain tumors receiving a standard dose of oral temozolomide displayed plasma and cerebrospinal fluid (CSF) TMZ concentrations of only up to about 70 µM and 10 µM, respectively [66, 67]. Similarly, patients treated for advanced neoplasms receiving combination chemotherapy comprising cyclophosphamide, cisplatin, and carmustine were found to have a mean peak plasma concentration of carmustine of 7.8 µM [68]. The development of implantable carmustine wafers allowed drug concentrations of up to several hundred times the concentration achievable with intravenously administered doses in a brain tumor environment [69].
The C6 glioma cell line was chosen for this study because the cell line exhibits high CD44 expression [70]. Moreover, most of the C6 cells are cancer stem-like cells with in vitro characteristics of self-renewal [71–72]. That makes this cell line particularly suitable to study the reversal of resistance to anti-cancer drugs [61]. However, as cancer cell line models are continuously questioned [73], the possible clinical relevance of the obtained results requires further investigations.

The authors declare no conflict of interests.
The study was supported by the Medical University of Lodz with the grant for young scientists No. 502-03/1-023-01/502-14-159.

References

1. Lippert TH, Ruoff HJ, Volm M. Current status of methods to assess cancer drug resistance. Int J Med Sci 2011; 8: 245-53.
2. Liu FS. Mechanisms of chemotherapeutic drug resistance in cancer therapy – a quick review. Taiwan J Obstet Gynecol 2009; 48: 239-44.
3. Gottesman MM. Mechanisms of cancer drug resistance. Annu Rev Med 2002; 53: 615-27.
4. Huang Y. Pharmacogenetics/genomics of membrane transporters in cancer chemotherapy. Cancer Metastasis Rev 2007; 26: 183-201.
5. Damaraju VL, Damaraju S, Young JD, Baldwin SA, Mackey J, Sawyer MB, Cass CE. Nucleoside anticancer drugs: the role of nucleoside transporters in resistance to cancer chemotherapy. Oncogene 2003; 22: 7524-36.
6. Sun YL, Patel A, Kumar P, Chen ZS. Role of ABC transporters in cancer chemotherapy. Chin J Cancer 2012; 31: 51-7.
7. Nobili S, Landini I, Mazzei T, Mini E. Overcoming tumor multidrug resistance using drugs able to evade P-glycoprotein or to exploit its expression. Med Res Rev 2012; 32: 1220-62.
8. Bardenheuer W, Lehmberg K, Rattmann I, et al. Resistance to cytarabine and gemcitabine and in vitro selection of transduced cells after retroviral expression of cytidine deaminase in human hematopoietic progenitor cells. Leukemia 2005;19: 2281-8.
9. Kasahara K, Fujiwara Y, Nishio K, Ohmori T, Sugimoto Y, Komiya K, Matsuda T, Saijo N. Metallothionein content correlates with the sensitivity of human small cell lung cancer cell lines to cisplatin. Cancer Res 1991; 51: 3237-42.
10. Rochat B. Role of cytochrome P450 activity in the fate of anticancer agents and in drug resistance: focus on tamoxifen, paclitaxel and imatinib metabolism. Clin Pharmacokinet 2005; 44: 349-66.
11. Bean J, Riely GJ, Balak M, Marks JL, Ladanyi M, Miller VA, Pao W. Acquired resistance to epidermal growth factor receptor kinase inhibitors associated with a novel T854A mutation in a patient with EGFR-mutant lung adenocarcinoma. Clin Cancer Res 2008; 14: 7519-25.
12. Apperley JF. Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol 2007; 8: 1018-29.
13. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007; 316: 1039-43.
14. Jiang G, Li LT, Xin Y, Zhang L, Liu YQ, Zheng JN. Strategies to improve the killing of tumors using temozolomide: targeting the DNA repair protein MGMT. Curr Med Chem 2012; 19: 3886-92.
15. Wilson TR, Johnston PG, Longley DB. Anti-apoptotic mechanisms of drug resistance in cancer. Curr Cancer Drug Targets 2009; 9: 307-19.
16. Reed JC. Apoptosis mechanisms: implications for cancer drug discovery. Oncology (Williston Park) 2004; 18: 11-20.
17. Wysocki PJ, Korski K, Lamperska K, Zaluski J, Mackiewicz A. Primary resistance to docetaxel-based chemotherapy in metastatic breast cancer patients correlates with a high frequency of BRCA1 mutations. Med Sci Monit 2008; 14: SC7-10.
18. Zahreddine H, Borden KL. Mechanisms and insights into drug resistance in cancer. Front Pharmacol 2013; 4: 28.
19. Meads MB, Gatenby RA, Dalton WS. Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat Rev Cancer 2009; 9: 665-74.
20. Correia AL, Bissell MJ. The tumor microenvironment is a dominant force in multidrug resistance. Drug Resist Updat 2012; 15: 39-49.
21. Frassanito MA, Cusmai A, Iodice G, Dammacco F. Autocrine interleukin-6 production and highly malignant multiple myeloma: relation with resistance to drug-induced apoptosis. Blood 2001; 97: 483-9.
22. Voorhees PM, Chen Q, Kuhn DJ, et al. Inhibition of interleukin-6 signaling with CNTO 328 enhances the activity of bortezomib in preclinical models of multiple myeloma. Clin Cancer Res 2007; 13: 6469-78.
23. Hazlehurst LA, Argilagos RF, Dalton WS. Beta1 integrin mediated adhesion increases Bim protein degradation and contributes to drug resistance in leukaemia cells. Br J Haematol 2007; 136: 269-75.
24. Toole BP, Slomiany MG. Hyaluronan: a constitutive regulator of chemoresistance and malignancy in cancer cells. Semin Cancer Biol 2008; 18: 244-50.
25. Laurent TC. Biochemistry of hyaluronan. Acta Otolaryngol Suppl 1987; 442: 7-24.
26. Laurent TC, Fraser JR. Hyaluronan. FASEB J 1992; 6: 2397-404.
27. Fraser JR, Laurent TC, Laurent UB. Hyaluronan: its nature, distribution, functions and turnover. J Intern Med 1997; 242: 27-33.
28. Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer 2013; 108: 1-8.
29. Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 2013; 62: 112-20.
30. Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer 2004; 4: 528-39.
31. Vabres P. Hyaluronan, embryogenesis and morphogenesis. Ann Dermatol Venereol 2010; 137 Suppl 1: S9-S14.
32. Bollyky PL, Bogdani M, Bollyky JB, Hull RL, Wight TN. The role of hyaluronan and the extracellular matrix in islet inflammation and immune regulation. Curr Diab Rep 2012; 12: 471-80.
33. Olczyk P, Komosińska-Vassev K, Winsz-Szczotka K, Kuźnik-Trocha K, Olczyk K. Hyaluronan: structure, metabolism, functions, and role in wound healing. Postepy Hig Med Dosw (Online) 2008; 62: 651-9.
34. Karbownik MS, Nowak JZ. Hyaluronan in cancer – pathophysiology and pharmacotherapy perspectives. J Oncol 2011; 61: 380-95.
35. Negi LM, Talegaonkar S, Jaggi M, Ahmad FJ, Iqbal Z, Khar RK. Role of CD44 in tumour progression and strategies for targeting. J Drug Target 2012; 20: 561-73.
36. Misra S, Heldin P, Hascall VC, Karamanos NK, Skandalis SS, Markwald RR, Ghatak S. Hyaluronan-CD44 interactions as potential targets for cancer therapy. FEBS J 2011; 278: 1429-43.
37. Karbownik MS, Nowak JZ. Hyaluronan: towards novel anti-cancer therapeutics. Pharmacol Rep 2013; 65: 1056-74.
38. Xu Y, Stamenkovic I, Yu Q. CD44 attenuates activation of the hippo signaling pathway and is a prime therapeutic target for glioblastoma. Cancer Res 2010; 70: 2455-64.
39. da Cunha CB, Oliveira C, Wen X, et al. De novo expression of CD44 variants in sporadic and hereditary gastric cancer. Lab Invest 2010; 90: 1604-14.
40. Desai B, Ma T, Zhu J, Chellaiah MA. Characterization of the expression of variant and standard CD44 in prostate cancer cells: identification of the possible molecular mechanism of CD44/MMP9 complex formation on the cell surface. J Cell Biochem 2009; 108: 272-84.
41. Wang SJ, Wong G, de Heer AM, Xia W, Bourguignon LY. CD44 variant isoforms in head and neck squamous cell carcinoma progression. Laryngoscope 2009; 119: 1518-30.
42. Liu CM, Chang CH, Yu CH, Hsu CC, Huang LL. Hyaluronan substratum induces multidrug resistance in human mesenchymal stem cells via CD44 signaling. Cell Tissue Res 2009; 336: 465-75.
43. Torre C, Wang SJ, Xia W, Bourguignon LY. Reduction of hyaluronan-CD44-mediated growth, migration, and cisplatin resistance in head and neck cancer due to inhibition of Rho kinase and PI-3 kinase signaling. Arch Otolaryngol Head Neck Surg 2010; 136: 493-501.
44. Wang SJ, Bourguignon LY. Role of hyaluronan-mediated CD44 signaling in head and neck squamous cell carcinoma progression and chemoresistance. Am J Pathol 2011; 178: 956-63.
45. Bourguignon LY, Earle C, Wong G, Spevak CC, Krueger K. Stem cell marker (Nanog) and Stat-3 signaling promote MicroRNA-21 expression and chemoresistance in hyaluronan/CD44-activated head and neck squamous cell carcinoma cells. Oncogene 2012; 31: 149-60.
46. Misra S, Ghatak S, Zoltan-Jones A, Toole BP. Regulation of multidrug resistance in cancer cells by hyaluronan. J Biol Chem 2003; 278: 25285-8.
47. Ghatak S, Misra S, Toole BP. Hyaluronan oligosaccharides inhibit anchorage-independent growth of tumor cells by suppressing the phosphoinositide 3-kinase/Akt cell survival pathway. J Biol Chem 2002; 277: 38013-20.
48. Misra S, Ghatak S, Toole BP. Regulation of MDR1 expression and drug resistance by a positive feedback loop involving hyaluronan, phosphoinositide 3-kinase, and ErbB2. J Biol Chem 2005; 280: 20310-5.
49. Ohashi R, Takahashi F, Cui R, et al. Interaction between CD44 and hyaluronate induces chemoresistance in non-small cell lung cancer cell. Cancer Lett 2007; 252: 225-34.
50. Gilg AG, Tye SL, Tolliver LB, et al. Targeting hyaluronan interactions in malignant gliomas and their drug-resistant multipotent progenitors. Clin Cancer Res 2008; 14: 1804-13.
51. Bacso Z, Nagy H, Goda K, Bene L, Fenyvesi F, Matkó J, Szabó G. Raft and cytoskeleton associations of an ABC transporter: P-glycoprotein. Cytometry A 2004; 61: 105-16.
52. Coley HM. Overcoming multidrug resistance in cancer: clinical studies of p-glycoprotein inhibitors. Methods Mol Biol 2010; 596: 341-58.
53. Baird RD, Kaye SB. Drug resistance reversal – are we getting closer? Eur J Cancer 2003; 39: 2450-61.
54. Lompardía SL, Papademetrio DL, Mascaró M, Alvarez EM, Hajos SE. Human leukaemic cell lines synthesize hyaluronan to avoid senescence and resist chemotherapy. Glycobiology 2013; 12: 1463-76.
55. Qin Z, Dai L, Bratoeva M, Slomiany MG, Toole BP, Parsons C. Cooperative roles for emmprin and LYVE-1 in the regulation of chemoresistance for primary effusion lymphoma. Leukemia 2011; 25: 1598-609.
56. Slomiany MG, Dai L, Bomar PA, Knackstedt TJ, Kranc DA, Tolliver L, Maria BL, Toole BP. Abrogating drug resistance in malignant peripheral nerve sheath tumors by disrupting hyaluronan-CD44 interactions with small hyaluronan oligosaccharides. Cancer Res 2009; 69: 4992-8.
57. Cui X, Zhou S, Xu H, Zhao T, Liu A, Guo X, Wang F. Reversal effects of hyaluronan oligosaccharides on adriamycin resistance of K562/A02 cells. Anticancer Drugs. 2009; 20: 800-6.
58. Toole BP, Ghatak S, Misra S. Hyaluronan oligosaccharides as a potential anticancer therapeutic. Curr Pharm Biotechnol 2008; 9: 249-52.
59. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352: 987-96.
60. Brandes AA, Tosoni A, Franceschi E, Reni M, Gatta G, Vecht C. Glioblastoma in adults. Crit Rev Oncol Hematol 2008; 67: 139-52.
61. Murat A, Migliavacca E, Gorlia T, et al. Stem cell-related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol 2008; 26: 3015-24.
62. Brandes AA, Fiorentino MV. The role of chemotherapy in recurrent malignant gliomas: an overview. Cancer Invest 1996; 14: 551-9.
63. Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, Wick W. Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res 2008; 14: 2900-8.
64. Peigńan L, Garrido W, Segura R, Melo R, Rojas D, Cárcamo JG, San Martín R, Quezada C. Combined use of anticancer drugs and an inhibitor of multiple drug resistance-associated protein-1 increases sensitivity and decreases survival of glioblastoma multiforme cells in vitro. Neurochem Res 2011; 36: 1397-406.
65. Lesley J, Hascall VC, Tammi M, Hyman R. Hyaluronan binding by cell surface CD44. J Biol Chem 2000; 275: 26967-75.
66. Ostermann S, Csajka C, Buclin T, Leyvraz S, Lejeune F, Decosterd LA, Stupp R. Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients. Clin Cancer Res 2004; 10: 3728-36.
67. Portnow J, Badie B, Chen M, Liu A, Blanchard S, Synold TW. The neuropharmacokinetics of temozolomide in patients with resectable brain tumors: potential implications for the current approach to chemoradiation. Clin Cancer Res 2009; 15: 7092-8.
68. Henner WD, Peters WP, Eder JP, Antman K, Schnipper L, Frei E 3rd. Pharmacokinetics and immediate effects of high-dose carmustine in man. Cancer Treat Rep 1986; 70: 877-80.
69. Lin SH, Kleinberg LR. Carmustine wafers: localized delivery of chemotherapeutic agents in CNS malignancies. Expert Rev Anticancer Ther 2008; 8: 343-59.
70. Knüpfer MM, Poppenborg H, Hotfilder M, Domula M, Wolff JE. Hyaluronic acid binding capacity of malignant glioma cells. Anticancer Res 1998; 18: 353-6.
71. Zhou XD, Wang XY, Qu FJ, et al. Detection of cancer stem cells from the C6 glioma cell line. J Int Med Res 2009; 37: 503-10.
72. Shen G, Shen F, Shi Z, Liu W, Hu W, Zheng X, Wen L, Yang X. Identification of cancer stem-like cells in the C6 glioma cell line and the limitation of current identification methods. In Vitro Cell Dev Biol Anim 2008; 44: 280-9.
73. Gillet JP, Varma S, Gottesman MM. The clinical relevance of cancer cell lines. J Natl Cancer Inst 2013; 105: 452-8.

Address for correspondence
Michał Seweryn Karbownik

Department of Pharmacology and Toxicology
Medical University of Lodz
Żeligowskiego 7/9
90-752 Lodz, Poland
e-mail: michal.karbownik@umed.lodz.pl

Submitted: 23.10.2013
Accepted: 11.04.2014
Copyright: © 2014 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.